Skip to main content

Main menu

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • Peer reviewer login
  • Alerts
  • Subscriptions

First-line carboplatin plus pemetrexed with pemetrexed maintenance in HIV-positive patients with advanced non-squamous non-small cell lung cancer: the phase II IFCT-1001 CHIVA trial

Armelle Lavole, Laurent Greillier, Julien Mazières, Isabelle Monnet, Lize Kiakouama-Maleka, Xavier Quantin, Jean Philippe Spano, Herve Lena, Philippe Fraisse, Henri Janicot, Clarisse Audigier-Valette, Alexandra Langlais, Franck Morin, Alain Makinson, Jacques Cadranel on behalf of the French Cooperative Thoracic Intergroup (IFCT)
European Respiratory Journal 2020 56: 1902066; DOI: 10.1183/13993003.02066-2019
Armelle Lavole
1Service de Pneumologie, Assistance Publique – Hôpitaux de Paris (Hôpital Tenon) and Sorbonne Université, Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: armelle.lavole@aphp.fr
Laurent Greillier
2Service d'Oncologie Multidisciplinaire & Innovations Thérapeutiques, Aix-Marseille Université, Assistance Publique - Hôpitaux de Marseille (Hôpital Nord), Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Laurent Greillier
Julien Mazières
3Service de Pneumologie, CHU, Toulouse, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Isabelle Monnet
4Service de Pneumologie, CHI, Créteil, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lize Kiakouama-Maleka
5Service de Pneumologie, HCL – Hôpital de la Croix-Rousse, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xavier Quantin
6Service de Pneumologie, CHU, Montpellier, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jean Philippe Spano
7Service d'Oncologie Médicale, Assistance Publique – Hôpitaux de Paris (Hôpital de la Pitié-Salpêtrière), Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Herve Lena
8Service de Pneumologie, CHU, Rennes, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Philippe Fraisse
9Service de Pneumologie, Nouvel Hôpital Civil, Strasbourg, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Henri Janicot
10Service de Pneumologie, CHU, Clermont-Ferrand, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clarisse Audigier-Valette
11Service de Pneumologie, CHI, Toulon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexandra Langlais
12French Cooperative Thoracic Intergroup (IFCT), Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Franck Morin
12French Cooperative Thoracic Intergroup (IFCT), Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alain Makinson
13Service des maladies infectieuses et tropicales, CHU Montpellier and InsermU1175, Université de Montpellier, Montpellier, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Alain Makinson
Jacques Cadranel
1Service de Pneumologie, Assistance Publique – Hôpitaux de Paris (Hôpital Tenon) and Sorbonne Université, Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

HIV infection is an exclusion criterion in lung cancer trials. This multicentre phase II trial aimed to assess feasibility, efficacy and safety of first-line carboplatin plus pemetrexed (CaP) followed by pemetrexed (P) maintenance in people living with HIV (PLHIV) with advanced non-squamous non-small cell lung cancer (NS-NSCLC).

Four cycles of CaP were followed by P-maintenance therapy in patients with Eastern Cooperative Oncology Group performance status ≤2. The primary objective was a disease control rate (DCR) ≥30% after 12 weeks.

Of the 61 PLHIV enrolled, 49 (80%) had a performance status of 0–1, and 19 (31%) had brain metastases. Median CD4 lymphocyte count was 418 cells·µL−1 (range 18–1230), median CD4 lymphocyte nadir was 169.5 cells·µL−1 (1–822); 48 (80%) patients were virologically controlled. Four-cycle inductions were achieved by 38 (62%) patients, and 31 (51%) started P-maintenance (median of 4.1 cycles (range 1–19)). The 12-week DCR was 50.8% (95% CI 38.3–63.4) and partial response rate 21.3%. Median progression-free survival and overall survival were 3.5 (95% CI 2.7–4.4) and 7.6 months (5.7–12.8), respectively. Patients with a performance status of 0–1 had the longest median progression-free survival (4.3 months, 95% CI 3.1–5.2) and overall survival (11.9 months, 95% CI 6.4–14.3). During induction, CaP doublet was well tolerated apart from grade 3–4 haematological toxicities (neutropenia 53.8%; thrombocytopenia 35.0%; anaemia 30.0%). Two fatal treatment-related sepses were reported. No opportunistic infections were experienced.

In PLHIV with advanced NS-NSCLC, first-line four-cycle CaP induction followed by P-maintenance was effective and reasonably well-tolerated. Further studies should evaluate combination strategies of CaP with immunotherapy in PLHIV.

Abstract

In this first clinical trial dedicated to people living with HIV and advanced non-squamous non-small cell lung cancer, first-line 4-cycle carboplatin plus pemetrexed followed by pemetrexed maintenance chemotherapy was effective and reasonably well-tolerated https://bit.ly/2xAqeEl

Introduction

The use of highly active antiretroviral therapy (HAART) in developed countries led to a considerable reduction in AIDS mortality due to opportunistic infections and AIDS-defining cancers [1]. In people living with HIV (PLHIV), non-AIDS-related cancers have become a leading cause of morbidity and mortality, with lung cancer being the first cause of mortality by cancer [2–5]. PLHIV have a greater risk of lung cancer compared with the general population [4, 6, 7], with an estimated 52% (95% CI 43–60) excess in the USA in 2015 [8]. However, the high prevalence of smoking in PLHIV is the primary contributor to lung cancer high incidence [4, 9–11].

Classically, the prognosis of lung cancer is worse in PLHIV [12, 13], but a recent French study showed that lung cancer prognosis in immunologically well-controlled PLHIV was similar to the general population [14].

Different national or expert working group guidelines recommend that PLHIV be treated with the same strategies as in the general population, with close monitoring of interactions between chemotherapy and HAART [15–18]. No trial dedicated to PLHIV with non-squamous non-small cell lung cancer (NS-NSCLC) had been conducted at the time of our study design. Before the era of immunotherapy, first-line chemotherapy with a platinum agent followed by pemetrexed (P)-maintenance therapy represented the standard regimen in the general population with advanced NS-NSCLC in the absence of oncogenic addiction [19–21]. Carboplatin pemetrexed (CaP), with low toxicity in the general population, seemed an ideal candidate in PLHIV with advanced NS-NSCLC, particularly if HAART comprised tenofovir and chemotherapy cisplatin, as both have been incriminated in tubular nephrotoxicity [22]. Moreover, pemetrexed undergoes non-CYP450 metabolism, contrasting with taxanes or vinorelbine, and the CaP doublet is thus unlikely to be altered by HAART [17, 23]. Finally, CaP has shown efficacy in frail NSCLC populations [24–26].

Based on these presumptions, we initiated a phase II, multicentre, non-randomised, open-label clinical trial to evaluate the efficacy and toxicity of CaP as first-line chemotherapy in PLHIV with advanced NS-NSCLC.

Methods

Eligibility criteria

To be eligible, PLHIV were diagnosed with histological or cytological confirmed NS-NSCLC [27], tumour–node–metastasis (TNM) stage III–IV, and had no previous administration of chemotherapy [28]. Cytopathological diagnosis and molecular testing were performed in each clinical centre. Molecular testing included: 1) immunohistochemistry (IHC) for ALK, confirmed by fluorescence in situ hybridisation in case of IHC positivity; and 2) EGFR (exon 18–21), KRAS (exon 2 and 13) and BRAF V600 mutation testing using the Sanger sequencing method or a more sensitive, validated, allele-specific technique. Other eligibility criteria were an age between 18 and 75 years, an Eastern Cooperative Oncology Group performance status (ECOG-PS) 0–2, a weight loss >10% of total body weight within the previous month and normal organ functions. Finally, patients with symptomatic or asymptomatic brain metastases could be included without prior treatment. This clinical trial (ClinicalTrials.gov, number NCT01296113) was approved by an Ethic Committee, and complied with French legislation, Good Clinical Practices, and the principles outlined in the latest version of Declaration of Helsinki. All patients gave their written informed study-specific consent.

Chemotherapy regimen

Patients received first-line intravenous chemotherapy with pemetrexed, 500 mg·m−2, bolus infused in 10 min, every 3 weeks, combined with carboplatin bolus infusion, with a target area under the curve value of 5 mg·mL−1·min−1 [29], on day 1 for a maximum of four cycles. After the four-cycle induction, patients achieving disease control (partial responses or stable diseases) and ECOG-PS ≤2 were continued on pemetrexed. Folic acid and vitamin B12 supplementation were mandatory, and prednisone 20 mg twice daily for 3 days was routinely administered.

Concomitant therapy

No other anticancer therapies, immunotherapies, hormonal cancer therapies, therapeutic radiotherapy (palliative radiotherapy being authorised) or experimental medications were permitted while patients were on study trial. Chemoprophylaxis against Pneumocystis pneumonia (PCP) and toxoplasmosis with trimethoprim-sulfamethoxazole (TMP–SMX), or alternative agents (pentamidine, atovaquone) in case of allergy or intolerance, was left to the investigator's appreciation but recommended if CD4 count was or fell below 200 cells·µL−1 during follow up.

Study objectives

The primary objective was a disease control rate (DCR) of at least 30% at week 12. Secondary objectives were progression-free survival (PFS), overall survival; overall survival and PFS according to performance status in class (0–1 and 2), quality of life (QoL) and overall toxicity. As we hypothesised that PLHIV with lung cancer were more at risk of infectious complications under chemotherapy than persons without HIV [30], safety objectives included characterising chemotherapy doublet toxicities, HIV viral load and CD4 lymphocyte count changes as well as opportunistic infections.

Study end points

12-week DCR was defined as the aggregated rates of complete or partial responses and stable diseases. PFS was defined as the time between patient's inclusion and disease progression, relapse or death of any cause, and overall survival as the time between inclusion and death from any cause.

Response to treatment was classified according to the Response Evaluation Criteria in Solid Tumors (RECIST) criteria v1.1 [31]. Tumour assessments were made by physical examination and total body computed tomodensitometry scan, which included brain evaluation, every two cycles. Primary criterion was DCR evaluated within the 4 weeks following completion of the 4-cycle induction phase.

Toxicity was scored every 3 weeks during induction period and according to usual practice scheduled during maintenance period, using the National Cancer Institute Common Adverse Events Criteria (CTCAE), version 3.0 [32]. QoL was evaluated using the Lung Cancer Symptom Scale (LCSS) at each cycle [33].

Statistical considerations

All patients were included in an intention-to-treat analyses for efficacy. The safety population comprised all patients who received at least one cycle of study chemotherapy.

Using the Fleming one-step method, and assuming that 12-week DCR of 30% or less (null hypothesis) was of no therapeutic interest and that a target DCR of at least 50% (alternative hypothesis) defined the doublet clinically efficient, 62 eligible patients were required (with 95% power and one-sided α error of 0.05). A triangular test [34] was used to analyse non-haematological grade 3–4 toxicities occurring with first-line chemotherapy (cycle 1–4) [35, 36]. Thirty percent (p0) or less of patients with non-haematological grade 3–4 toxicities (except for nausea and vomiting) was considered reasonable in this population, and 50% (p1) or more considered unacceptable. Two safety runs (with one-sided α error of 0.05 and a power of 95%) were planned after 10 and 31 treated patients to stop the trial for non-haematological toxicity if more than eight or 15 patients with cases of grade ≥3 non-haematological toxicity (excluding nausea and vomiting) occurred between cycles 1 and 4 of chemotherapy respectively. PFS and overall survival were estimated using Kaplan–Meier method with a censure of the follow-up on June 30, 2017. Hazard ratios and 95% confidence intervals for overall survival and PFS were estimated using a Cox model to select the most promising prognostic variables. The variables to be tested in the multivariable model were selected based on univariate analysis results (p<0.20). In the multivariable model, a p<0.05 was considered statistically significant. The QoL was described by the sum of the score of all domains of LCSS questionnaire, at the end of each first-line chemotherapy cycle compared to the baseline value [37].

Statistical analyses were performed using SAS software version 9.4; all p-values and confidence intervals were two sided.

Results

Patients' characteristics

Between May 2011 and July 2015, 61 patients were enrolled (figure 1). Their baseline characteristics are summarised in table 1. The HIV viral load was undetectable (<50 copies·mL−1) in 48 of the 60 documented patients (80.0%), and median CD4 count was 418 cells·µL−1 (range 18–1230). At inclusion, 58 patients (95.1%) were treated (detailed in supplementary table A1), 12 (19.6%) were ECOG-PS2 and 19 (31.1%) presented with brain metastases, of whom nine were symptomatic.

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

CONSORT flow chart. CaP: carboplatin plus pemetrexed

View this table:
  • View inline
  • View popup
TABLE 1

Patients' characteristics at baseline

Chemotherapy administration

60 patients received at least one first-line CaP cycle. A dose reduction was reported for carboplatin and for pemetrexed in 17 patients (27.9%). The relative dose-intensities were 95.9% for pemetrexed and 86.1% for carboplatin. The full planned induction regimen (four cycles) was completed in 38 patients (62.3%). The remaining 22 patients (36.1%) withdrew because of disease progression (n=13), death (n=6), adverse events (n=2) or patient's choice (n=1). Among the 38 four-cycle completers, 31 patients (50.8% of efficacy population) started pemetrexed-maintenance (figure 1) and received a median of four maintenance cycles (range: 1–19) with a relative dose-intensity of 95.1%.

Disease response

The primary objective of 12-week DCR ≥30% was reached as 31 patients (50.8% (95% CI 38.3–63.4)) of the efficacy population had disease control while a minimum of 26 patients were hypothesised for treatment success (table 2). Objective response and stabilisation rates were 21.3 and 29.5%, respectively. Considering the patients with ECOG-PS 0–1, 57.1% of them reached disease control (versus 50.8% in overall population), reflecting an increase in the rate of stable disease (34.7% in ECOG-PS 0–1 versus 29.5% in the overall population).

View this table:
  • View inline
  • View popup
TABLE 2

Best overall response during the 4-cycle first-line chemotherapy

PFS and overall survival

The median follow-up duration of patients was 45.5 months (range 23.9–73.2). The median PFS was 3.5 months (95% CI 2.7–4.4) (figure 2a), and the median overall survival 7.6 months (95% CI 5.7–12.8) (figure 2b). After treatment discontinuation, 37 (60.7%) received second-line chemotherapy (supplementary table A2). 55 patients (90.2%) died, mostly due to NSCLC progression (47 out of 55 patients (85.5%)).

FIGURE 2
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2

Progression-free survival and overall survival (from inclusion): Kaplan Meier curves. a) progression-free survival. b) overall survival. OS: overall survival; PFS: progression-free survival.

Using a multivariable Cox's model, the only patient and disease characteristics at baseline significantly related to PFS and overall survival was the ECOG-PS (supplementary tables A3 and A4). PS 2 versus PS 0–1 patients had shorter PFS and overall survival (HR 2.67 (95% CI 1.31–5.43) and 4.44 (95% CI 1.93–10.22), respectively; data not shown). Exploratory analyses showed that patients with PS 0–1 had much longer median PFS and overall survival than PS-2 patients (figure 3). Patients without brain metastases status had longer median overall survival, with no difference in PFS (supplementary figure A1). Patients entering the pemetrexed-maintenance period had increased survival rates (PFS 5.6 months (95% CI 4.8–7.0); overall survival 16.9 months (95% CI 11.8–23.1) (supplementary figure A2).

FIGURE 3
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3

Progression-free survival and overall survival (from inclusion) according to Eastern Cooperative Oncology Group performance status (ECOG-PS) class at baseline: Kaplan Meier curves. a) Progression-free survival. b) Overall survival. OS: overall survival; PFS: progression-free survival.

Safety

Chemotherapy-related toxicity

During the 12-week induction period, grade 3 to 4 treatment-related adverse events occurring in at least 10% of patients were neutropenia (53.3%) thrombocytopenia (35.0%), anaemia, (30.0%) and asthenia (16.7%) (table 3). In the 58 patients treated with ART at inclusion, AIDS history was the only risk factor of grade 3–4 haematological toxicity (p=0.05; table 4). The safety runs on non-haematological grade 3–4 toxicities (except nausea and vomiting) did not lead to study discontinuation. No grade 3–4 renal toxicity was reported and, among the 4 grade 1–2 renal toxicity, one patient was treated with tenofovir. Two chemotherapy-related death were reported due to sepsis (two out of 61 (3%)). In the 31 patients treated during the pemetrexed-maintenance period, no treatment-related deaths were reported.

View this table:
  • View inline
  • View popup
TABLE 3

Grade 1 to 5 toxicity of the carboplatin-pemetrexed doublet

View this table:
  • View inline
  • View popup
TABLE 4

Risks factors of grade 3 to 4 haematologic toxicity of the carboplatin-pemetrexed doublet in patients treated with antiretroviral therapy at study enrolment (n=58)

Immunodeficiency-related adverse events

No opportunistic infections were reported. Nine patients (nine out of 12 (75%)) with TCD4 lymphocytes <200 cell·µL−1 received cotrimoxazole prophylaxis. 23 infectious events were experienced, including 13 respiratory infections, five infections of the implantable venous access device or pleural drain, and five infections in other sites.

Changes in CD4 lymphocyte count and HIV viral load

In the intention to treat population, the absolute CD4 counts significantly diminished between baseline and the end of the four-cycle first-line chemotherapy (median (range) from 418 cells·µL−1 (18–1230) to 292 cells·µL−1 (50–1607); p=0.04). The last median CD4 count was 319.5 cells·µL−1 (72–816). After four chemotherapy doublet cycles, only two patients (treated with HAART at study enrolment) had detectable viral load (from 397 at baseline to 4916 copies·mL−1 and from <50 to 1638 copies·mL−1). Of the 23 patients with undetectable baseline viral load and with values available after inclusion, 22 remained undetectable (95.6%).

Quality of life

LCSS Questionnaire scores improved from baseline with an increase of score in 25–29% of patients after the first cycle of chemotherapy, but 9–21% of patients experienced a decline in their QoL (supplementary figure A3).

Discussion

Whereas PLHIV are traditionally excluded from lung cancer trials, we report the results of the first trial on first-line chemotherapy in PLHIV with NS-NSCLC. Half of the patients achieved disease control at the end of the four-cycle first-line CaP doublet treatment reaching the primary study objective of at least 30% of DCR. Patients were relatively young (median age 52.9 years), nearly all smokers, mainly ECOG-PS 0–1 (80%), and with controlled HIV disease. These features are relatively consistent with a previous French cohort and with most PLHIV under HAART in France [23]. The study DCR was 51% which is close to that of NS-NSCLC patients of the general population treated with pemetrexed-based doublet in the PARAMOUNT trial (57%) [20]. Overall survival was 7.6 months in our study while overall survival in PLHIV with advanced NSCLC still remained poor ranging from 3 to 7 months in previous retrospective cohorts [23–26, 33].

Despite being virologically well controlled, survival of PLHIV in our study seemed shorter than previously observed in NS-NSCLC patients of the general population, treated with platinum-based doublet followed by pemetrexed maintenance [20, 21]. However, in these studies, patients were ECOG-PS 0–1 at baseline whereas in our study, PLHIV were ECOG-PS 0–2 at baseline. Furthermore, our ECOG-PS 0–1 subgroup almost reached similar median overall survival [20, 21]. As observed in the general population, and in PLHIV [24, 25] ECOG-PS was significantly related to PFS and overall survival.

Patients entering pemetrexed-maintenance received a median of four cycles (range 1–19), consistent with reports in the general population (PARAMOUNT study: four cycles (range 1–19); JMEN trial: five cycles (range 1–55)] [20, 21]. These patients reached similar or sometimes longer survival rates than in the general population treated with platinum-based doublet [20, 21]. During both study periods, relative dose-intensities remained stable in a vast majority of patients, underlying good tolerability of the regimen and its compatibility with antiretroviral therapies. Likewise during induction, the QoL after each CaP cycle remained stable for most of the studied patients, and even improved for more than 25% of them after their first induction cycle.

No unexpected toxicity was observed, reflecting the absence or a low level of interactions with HAART [17, 23]. Only one renal toxicity grade 1–2 was reported under tenofovir. Two treatment-related deaths (3%) were noted, lower than a study with 6 out of 13 (46%) haematological deaths when HAART was associated with cytotoxic chemotherapy metabolised by the CYP450 [23]. The haematological toxicities of this carboplatin doublet were relatively high compared with other studies in the general population [27, 29], but similar to another study with elderly people [28]. AIDS was the only risk factor found to be associated with the haematological toxicity. PLHIV may be facing decreased haematopoiesis, particularly in those with a history of advanced HIV disease, which may persist despite HAART [38], justifying in some cancers in PLHIV systematic administration of growth factors such as granulocyte-colony stimulating factor [16, 18, 39, 40].

The CD4 count and viral load remained stable during chemotherapy. No toxoplasmosis or PCP cases were experienced. Prophylaxis was recommended only for patients with CD4 count <200 cells·μL−1, thus for a minority of persons. Our study argues against systematic prophylaxis independent of CD4 lymphocyte count in PLHIV with lung cancer, in contrast with the recommendations of the French expert panel and European AIDS Clinical Society (EACS) recommendations [40, 41], but in accordance with the British HIV Association and the NCCN guidelines [16, 18]. However, further studies with more PLHIV are necessary to confirm these findings.

The trial suffers from a bias related to the potential heterogeneity of the PLHIV population; however, the option of a restricted population of patients without PS 2 and without symptomatic brain metastases could have affected the recruitment of this first trial dedicated PLHIV with NS-NSCLC. Patients with oncogenic-driven NSCLC were not excluded and three patients would have received tyrosine kinase inhibitors as first-line treatments in the present era. Moreover, Anti-programmed death (PD)1 and anti-PD ligand (PDL)1 treatment regimens (immuno-oncology) are now the standard of care for non-oncogenic driven advanced NSCLC but at the time of the trial accrual (2011–2015) no data existed concerning the immuno-oncology therapeutic strategy and, therefore, the knowledge of programmed death ligand 1 (PD-L1) tumour was not mandatory. The treatment landscape of NSCLC has changed since the advent of immuno-oncology and trials are in progress in PLHIV with lung cancer (NTC03354936, NCT03304093). Real-life safety and efficacy data of immuno-oncology following chemotherapy in PLHIV with lung cancer and other cancers have been reported [43–45].

Recent guidelines recommend for patients with high (PD-L1) expression (tumour proportion score (TPS) ≥50%) and NS-NSCLC a single-agent pembrolizumab and for patients with either negative (0%) or low positive (1% to 49%) PD-L1 a pembrolizumab/platin/pemetrexed combination, although PLHIV were excluded from all these studies [42]. Our study will be a reference when assessing the potential impact on toxicities of a platin doublet in combination with immuno-oncology as first-line treatment for PLHIV with NS-NCLC.

In conclusion, this study conducted in PLHIV with NS-NSCLC showed the effectiveness of the induction doublet carboplatin/pemetrexed. Despite its relative haematological toxicity, it was well tolerated, without severe renal toxicity nor treatment-related unexpected deaths. This doublet is thus a relevant first-line chemotherapy in PLHIV with NS-NSCLC, leading to relatively long survivals, particularly in those entering pemetrexed maintenance. There is a crucial need for additional data on responses and tolerance in PLHIV of lung cancer therapies developed in the general population, such as immunotherapies [46].

Supplementary material

Supplementary Material

Please note: supplementary material is not edited by the Editorial Office, and is uploaded as it has been supplied by the author.

Supplementary table A1. Antiretroviral treatments at study enrolment. ERJ-02066-2019.Table_A1

Supplementary table A2. First post-study treatment. ERJ-02066-2019.Table_A2

Supplementary table A3. Prognosis analysis of progression-free survival. ERJ-02066-2019.Table_A3

Supplementary table A4. Prognosis analysis of overall survival. ERJ-02066-2019.Table_A4

Supplementary figure A1. Progression-free survival and overall survival (from inclusion) according to brain metastasis status at baseline. ERJ-02066-2019.Figure_A1

Supplementary figure A2. Progression-free survival and overall survival (from inclusion) of patients entering pemetrexed maintenance period. ERJ-02066-2019.Figure_A2

Supplementary figure A3. Changes in the mean score of global quality of life. ERJ-02066-2019.Figure_A3

Shareable PDF

Supplementary Material

This one-page PDF can be shared freely online.

Shareable PDF ERJ-02066-2019.Shareable

Acknowledgements

We thank the participating patients and their families as well as the study teams involved in the trial, the clinical research assistants, study coordinators and IFCT operations staff, as well as G. Cremer at Cremer Consulting (Strasbourg, France) for medical writing support. We thank also the French National Cancer Institute (INCa) and the French League against Cancer. Prior presentation: Presented at the 2016 annual ASCO meeting, Chicago, IL, USA, June 4, 2016.

CHIVA trial investigators: Armelle Lavole and Jacques Cadranel (Paris, AP-HP Hopital Tenon), Laurent Greillier and Fabrice Barlesi (Marseille, AP-HM), Julien Mazieres and Christophe Hermant (Toulouse, CHU), Isabelle Monnet (Créteil, CHI), Lize Kiakouama-Maleka (Lyon, HCL Hopital Croix-Rousse), Xavier Quantin (Montpellier, CHU), Jean-Philippe Spano and Laurent Taillade (Paris, AP-HP Hopital Pitié-Salpêtrière), Hervé Lena (Rennes, CHU), Philippe Fraisse (Strasbourg, Nouvel Hopital Civil), Henri Janicot (Clermont-Ferrand, CHU), Clarisse Audigier-Valette (Toulon, CHI), Gilles Pichancourt (Avignon, CH), Sophie Schneider (Bayonne, CH), Virginie Westeel (Besançon, CHU), Emmanuel Bergot (Caen, CHU), Patricia Barre (Cahors, CH), Lionel Moreau (Colmar, CH), Olivier Molinier (Le Mans, CHG), Dominique Herman (Nevers, CH), Aldo Renault (Pau, CHG), Hélène Doubre (Suresnes, Hopital Foch), Emilie Pluquet (Tourcoing, CH).

Footnotes

  • This article has supplementary material available from erj.ersjournals.com

  • Author contributions: Conception and design: A. Lavolé, L. Greiller, J. Mazières, I. Monnet, L. Kiakouama-Maleka, X. Quantin, J.P. Spano, H. Lena, P. Fraisse, H. Janicot, C. Audigier-Valette, A. Langlais, F. Morin, A. Makinson and J. Cadranel; financial support: F. Morin; administrative support: F. Morin; provision of study material or patients: A. Lavolé, L. Greiller, J. Mazières, I. Monnet, L. Kiakouama-Maleka, X. Quantin, J.P. Spano, H. Lena, P. Fraisse, H. Janicot, C. Audigier-Valette and J. Cadranel; collection and assembly of data: A. Lavolé, A. Langlais, F. Morin, A. Makinson and J. Cadranel; data analysis and interpretation: A. Lavolé, J. Mazières, A. Langlais, F. Morin, A. Makinson and J. Cadranel; manuscript writing: all authors; final approval of manuscript: all authors; accountable for all aspects of the work: all authors.

  • Conflict of interest: A. Lavole has nothing to disclose.

  • Conflict of interest: L. Greillier reports personal fees from AstraZeneca, Boehringer Ingelheim, Roche, Bristol-Myers Squibb, MSD, Takeda, Abbvie and Novartis, outside the submitted work; travel and accommodation expenses from MSD and Pfizer.

  • Conflict of interest: J. Mazières has nothing to disclose.

  • Conflict of interest: I. Monnet has nothing to disclose.

  • Conflict of interest: L. Kiakouama-Maleka has nothing to disclose.

  • Conflict of interest: X. Quantin has nothing to disclose.

  • Conflict of interest: J.P. Spano reports personal fees for consultancy from MSD, Roche and Biogaran, personal fees for advisory board work from Lilly, Mylan and Novartis, personal fees for advisory board work and lectures from Pfizer and Leopharma, personal fees for lectures from Pierre Fabre Oncology, AstraZeneca and Gilead, grants from BMS and MSD Avenir, outside the submitted work.

  • Conflict of interest: H. Lena has nothing to disclose.

  • Conflict of interest: P. Fraisse has nothing to disclose.

  • Conflict of interest: H. Janicot has nothing to disclose.

  • Conflict of interest: C. Audigier-Valette has nothing to disclose.

  • Conflict of interest: A. Langlais has nothing to disclose.

  • Conflict of interest: F. Morin has nothing to disclose.

  • Conflict of interest: A. Makinson has nothing to disclose.

  • Conflict of interest: J. Cadranel reports grants from AstraZeneca, Novartis and Pfizer, outside the submitted work.

  • Support statement: Lilly (France) supplied pemetrexed and a research grant to Intergroupe Francophone de Cancérologie Thoracique (IFCT) for the IFCT-1001 CHIVA Trial. Funding information for this article has been deposited with the Crossref Funder Registry.

  • Received October 22, 2019.
  • Accepted April 15, 2020.
  • Copyright ©ERS 2020
https://www.ersjournals.com/user-licence

References

  1. ↵
    1. Panel on Antiretroviral Guidelines for Adults and Adolescents
    . Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV Department of Health and Human Services. 2018. www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf
  2. ↵
    1. Silverberg MJ,
    2. Lau B,
    3. Achenbach CJ, et al.
    Cumulative incidence of cancer among persons with HIV in North America: A cohort study. Ann Intern Med 2015; 163: 507–518. doi:10.7326/M14-2768
    OpenUrlCrossRefPubMed
    1. Morlat P,
    2. Roussillon C,
    3. Henard S, et al.
    Causes of death among HIV-infected patients in France in 2010 (national survey): trends since 2000. AIDS 2014; 28: 1181–1191. doi:10.1097/QAD.0000000000000222
    OpenUrlCrossRefPubMedWeb of Science
  3. ↵
    1. Mani D,
    2. Haigentz M, Jr.,
    3. Aboulafia DM
    . Lung cancer in HIV infection. Clin Lung Cancer 2012; 13: 6–13. doi:10.1016/j.cllc.2011.05.005
    OpenUrlCrossRefPubMed
  4. ↵
    1. Engels EA,
    2. Yanik EL,
    3. Wheeler W, et al.
    Cancer-attributable mortality among people with treated human immunodeficiency virus infection in North America. Clin Infect Dis 2017; 65: 636–643. doi:10.1093/cid/cix392
    OpenUrl
  5. ↵
    1. Guiguet M,
    2. Boué F,
    3. Cadranel J, et al.
    Effect of immunodeficiency, HIV viral load, and antiretroviral therapy on the risk of individual malignancies (FHDH-ANRS CO4): a prospective cohort study. Lancet Oncol 2009; 10: 1152–1159. doi:10.1016/S1470-2045(09)70282-7
    OpenUrlCrossRefPubMedWeb of Science
  6. ↵
    1. Sigel K,
    2. Wisnivesky J,
    3. Gordon K, et al.
    HIV as an independent risk factor for incident lung cancer. AIDS 2012; 26: 1017–1025. doi:10.1097/QAD.0b013e328352d1ad
    OpenUrlCrossRefPubMedWeb of Science
  7. ↵
    1. Robbins HA,
    2. Pfeiffer RM,
    3. Shiels MS, et al.
    Excess cancers among HIV-infected people in the United States. J Natl Cancer Inst 2015; 107: dju503. doi:10.1093/jnci/dju503
    OpenUrlCrossRefPubMed
  8. ↵
    1. Winstone TA,
    2. Man SFP,
    3. Hull M, et al.
    Epidemic of lung cancer in patients with HIV infection. Chest 2013; 143: 305–314. doi:10.1378/chest.12-1699
    OpenUrlCrossRefPubMed
    1. Tron L,
    2. Lert F,
    3. Spire B, et al.
    Tobacco smoking in HIV-infected versus general population in France: heterogeneity across the various groups of people living with HIV. PLoS One 2014; 9: e107451. doi:10.1371/journal.pone.0107451
    OpenUrlCrossRefPubMed
  9. ↵
    1. Helleberg M,
    2. Gerstoft J,
    3. Afzal S, et al.
    Risk of cancer among HIV-infected individuals compared to the background population: impact of smoking and HIV. AIDS 2014; 28: 1499–1508. doi:10.1097/QAD.0000000000000283
    OpenUrlCrossRefPubMedWeb of Science
  10. ↵
    1. Suneja G,
    2. Shiels MS,
    3. Melville SK, et al.
    Disparities in the treatment and outcomes of lung cancer among HIV-infected individuals. AIDS 2013; 27: 459–468. doi:10.1097/QAD.0b013e32835ad56e
    OpenUrlCrossRefPubMed
  11. ↵
    1. Coghill AE,
    2. Shiels MS,
    3. Suneja G, et al.
    Elevated cancer-specific mortality among HIV-infected patients in the United States. J Clin Oncol 2015; 33: 2376–2383. doi:10.1200/JCO.2014.59.5967
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Hleyhel M,
    2. Belot A,
    3. Bouvier AM, et al.
    Trends in survival after cancer diagnosis among HIV-infected individuals between 1992 and 2009. Results from the FHDH-ANRSCO4 cohort. Int J Cancer 2015; 137: 2443–2453. doi:10.1002/ijc.29603
    OpenUrlCrossRefPubMed
  13. ↵
    1. Spano JP,
    2. Poizot-Martin I,
    3. Costagliola D, et al.
    Non-AIDS-related malignancies: expert consensus review and practical applications from the multidisciplinary CANCERVIH Working Group. Ann Oncol 2016; 27: 397–408. doi:10.1093/annonc/mdv606
    OpenUrlCrossRefPubMed
  14. ↵
    1. Bower M,
    2. Palfreeman A,
    3. Alfa-Wali M, et al.
    British HIV Association guidelines for HIV-associated malignancies 2014. HIV Med 2014; 15: Suppl. 2, 1–92.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Rudek MA,
    2. Flexner C,
    3. Ambinder RF
    . Use of antineoplastic agents in patients with cancer who have HIV/AIDS. Lancet Oncol 2011; 12: 905–912. doi:10.1016/S1470-2045(11)70056-0
    OpenUrlCrossRefPubMedWeb of Science
  16. ↵
    Cancer in people living with HIV, Version 1.2018. Clinical practice guidelines in oncology. J Natl Compr Canc Netw 2018; 16: 986–1017. doi:10.6004/jnccn.2018.0066
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Abernethy AP,
    2. Arunachalam A,
    3. Burke T, et al.
    Real-world first-line treatment and overall survival in non-small cell lung cancer without known EGFR mutations or ALK rearrangements in US community oncology setting. PLoS One 2017; 12: e0178420. doi:10.1371/journal.pone.0178420
    OpenUrlPubMed
  18. ↵
    1. Paz-Ares L,
    2. de Marinis F,
    3. Dediu M, et al.
    Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced nonsquamous non-small-cell lung cancer (PARAMOUNT): a double-blind, phase 3, randomised controlled trial. Lancet Oncol 2012; 13: 247–255. doi:10.1016/S1470-2045(12)70063-3
    OpenUrlCrossRefPubMedWeb of Science
  19. ↵
    1. Ciuleanu T,
    2. Brodowicz T,
    3. Zielinski C, et al.
    Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet 2009; 374: 1432–1440. doi:10.1016/S0140-6736(09)61497-5
    OpenUrlCrossRefPubMedWeb of Science
  20. ↵
    1. Ustianowski A,
    2. Arends JE
    . Tenofovir: what we have learnt after 7.5 million person-years of use. Infect Dis Ther 2015; 4: 145–157. doi:10.1007/s40121-015-0070-1
    OpenUrl
  21. ↵
    1. Makinson A,
    2. Tenon JC,
    3. Eymard-Duvernay S, et al.
    Human immunodeficiency virus infection and non-small cell lung cancer: survival and toxicity of antineoplastic chemotherapy in a cohort study. J Thorac Oncol 2011; 6: 1022–1029. doi:10.1097/JTO.0b013e318217b6e0
    OpenUrlCrossRefPubMed
  22. ↵
    1. Pakkala S,
    2. Ramalingam SS
    . Lung cancer in HIV-positive patients. J Thorac Oncol 2010; 5: 1864–1871. doi:10.1097/JTO.0b013e3181f387fd
    OpenUrlCrossRefPubMed
  23. ↵
    1. Lavolé A,
    2. Chouaïd C,
    3. Baudrin L, et al.
    Effect of highly active antiretroviral therapy on survival of HIV infected patients with non-small-cell lung cancer. Lung Cancer 2009; 65: 345–350. doi:10.1016/j.lungcan.2008.11.018
    OpenUrlCrossRefPubMedWeb of Science
  24. ↵
    1. Tirelli U,
    2. Spina M,
    3. Sandri S, et al.
    Lung carcinoma in 36 patients with human immunodeficiency virus infection. The Italian Cooperative Group on AIDS and Tumors. Cancer 2000; 88: 563–569. doi:10.1002/(SICI)1097-0142(20000201)88:3<563::AID-CNCR11>3.0.CO;2-D
    OpenUrlCrossRefPubMedWeb of Science
  25. ↵
    1. Zukin M,
    2. Barrios CH,
    3. Pereira JR, et al.
    Randomized phase III trial of single-agent pemetrexed versus carboplatin and pemetrexed in patients with advanced non-small-cell lung cancer and Eastern Cooperative Oncology Group performance status of 2. J Clin Oncol 2013; 31: 2849–2853. doi:10.1200/JCO.2012.48.1911
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Tamiya M,
    2. Tamiya A,
    3. Kaneda H, et al.
    A phase II study of pemetrexed plus carboplatin followed by maintenance pemetrexed as first-line chemotherapy for elderly patients with advanced non-squamous non-small cell lung cancer. Med Oncol 2016; 33: 2. doi:10.1007/s12032-015-0715-7
    OpenUrl
  27. ↵
    1. Zhao X,
    2. Yu H,
    3. Zhao J, et al.
    Efficacy and safety of first-line pemetrexed plus carboplatin followed by single-agent pemetrexed maintenance in elderly Chinese patients with non-squamous non-small-cell lung cancer. Oncotarget 2017; 23: 86384–86394. doi:10.18632/oncotarget.21186
    OpenUrl
  28. ↵
    1. Travis WD,
    2. Brambilla E,
    3. Nicholson AG, et al.
    The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol 2015; 10: 1243–1260. doi:10.1097/JTO.0000000000000630
    OpenUrlCrossRefPubMed
  29. ↵
    1. Edge SB,
    2. Compton CC
    . The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 2010; 17: 1471–1474. doi:10.1245/s10434-010-0985-4
    OpenUrlCrossRefPubMedWeb of Science
  30. ↵
    1. Calvert AH,
    2. Newell DR,
    3. Gumbrell LA, et al.
    Carboplatin dosage: prospective evaluation of a simple formula based on renal function. J Clin Oncol 1989; 7: 1748–1756. doi:10.1200/JCO.1989.7.11.1748
    OpenUrlAbstract
  31. ↵
    1. Brock MV,
    2. Hooker CM,
    3. Engels EA, et al.
    Delayed diagnosis and elevated mortality in an urban population with HIV and lung cancer: implications for patient care. J Acquir Immune Defic Syndr 2006; 43: 47–55. doi:10.1097/01.qai.0000232260.95288.93
    OpenUrlCrossRefPubMedWeb of Science
  32. ↵
    1. Bellissant E,
    2. Bénichou J,
    3. Chastang C
    . The group sequential triangular test for phase II cancer clinical trials. Am J Clin Oncol 1996; 19: 422–430. doi:10.1097/00000421-199608000-00021
    OpenUrlCrossRefPubMed
  33. ↵
    1. Therasse P,
    2. Arbuck SG,
    3. Eisenhauer EA, et al.
    New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92: 205–216. doi:10.1093/jnci/92.3.205
    OpenUrlCrossRefPubMedWeb of Science
  34. ↵
    1. Trotti A,
    2. Colevas AD,
    3. Setser A, et al.
    CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol 2003; 13: 176–181. doi:10.1016/S1053-4296(03)00031-6
    OpenUrlCrossRefPubMedWeb of Science
  35. ↵
    1. Hollen PJ,
    2. Gralla RJ,
    3. Kris MG, et al.
    Quality of life assessment in individuals with lung cancer: Testing the Lung Cancer Symptom Scale (LCSS). Eur J Cancer 1993; 29A: Suppl. 1, S51–S58. doi:10.1016/S0959-8049(05)80262-X
    OpenUrl
  36. ↵
    1. Isgrò A,
    2. Mezzaroma I,
    3. Aiuti A, et al.
    Recovery of hematopoietic activity in bone marrow from human immunodeficiency virus type 1-infected patients during highly active antiretroviral therapy. AIDS Res Hum Retroviruses 2000; 16: 1471–1479. doi:10.1089/088922200750005994
    OpenUrlCrossRefPubMedWeb of Science
  37. ↵
    1. Levine AM,
    2. Karim R,
    3. Mack W, et al.
    Neutropenia in human immunodeficiency virus infection: data from the women's interagency HIV study. Arch Intern Med 2006; 166: 405–410.
    OpenUrlCrossRefPubMedWeb of Science
  38. ↵
    Prise en charge médicale des personnes vivant avec le VIH Cancers (août 2017). Groupe d'experts pour la prise en charge du VIH. https://cns.sante.fr/wp-content/uploads/2017/10/experts-vih_cancers.pdf.
  39. ↵
    EACS Treatment Guidelines 10.0. 2020. www.eacsociety.org/files/2020_guidelines-10.0-english.pdf.
  40. ↵
    1. Hanna NH,
    2. Schneider BJ,
    3. Temin S
    . Therapy for stage IV non-small-cell lung cancer without driver alterations: ASCO and OH (CCO) Joint Guideline Update. J Clin Oncol 2020; 38: 1608–1632.
    OpenUrl
  41. ↵
    1. Cook MR,
    2. Kim C
    . Safety and efficacy of immune checkpoint inhibitor therapy in patients with HIV infection and advanced-stage cancer: a systematic review. JAMA Oncol 2019; 5: 1049–1054. doi:10.1001/jamaoncol.2018.6737
    OpenUrlPubMed
    1. Spano JP,
    2. Veyri M,
    3. Gobert A, et al.
    Immunotherapy for cancer in people living with HIV: safety with an efficacy signal from the series in real life experience. AIDS 2019; 33: F13–F19. doi:10.1097/QAD.0000000000002298
    OpenUrl
  42. ↵
    1. Ostios-Garcia L,
    2. Faig J,
    3. Giulia C, et al.
    Safety and efficacy of PD-1 inhibitors among hiv-positive patients with non-small cell lung cancer. J Thorac Oncol 2018; 13: 1037–1042. doi:10.1016/j.jtho.2018.03.031
    OpenUrl
  43. ↵
    Immunotherapy by nivolumab after prior chemotherapy for HIV+ patients with advanced non-small cell lung cancer (NSCLC): IFCT-CHIVA2 Phase IIa Trial. https://clinicaltrials.gov/ct2/show/NCT03304093
PreviousNext
Back to top
View this article with LENS
Vol 56 Issue 2 Table of Contents
European Respiratory Journal: 56 (2)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
First-line carboplatin plus pemetrexed with pemetrexed maintenance in HIV-positive patients with advanced non-squamous non-small cell lung cancer: the phase II IFCT-1001 CHIVA trial
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
First-line carboplatin plus pemetrexed with pemetrexed maintenance in HIV-positive patients with advanced non-squamous non-small cell lung cancer: the phase II IFCT-1001 CHIVA trial
Armelle Lavole, Laurent Greillier, Julien Mazières, Isabelle Monnet, Lize Kiakouama-Maleka, Xavier Quantin, Jean Philippe Spano, Herve Lena, Philippe Fraisse, Henri Janicot, Clarisse Audigier-Valette, Alexandra Langlais, Franck Morin, Alain Makinson, Jacques Cadranel
European Respiratory Journal Aug 2020, 56 (2) 1902066; DOI: 10.1183/13993003.02066-2019

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
First-line carboplatin plus pemetrexed with pemetrexed maintenance in HIV-positive patients with advanced non-squamous non-small cell lung cancer: the phase II IFCT-1001 CHIVA trial
Armelle Lavole, Laurent Greillier, Julien Mazières, Isabelle Monnet, Lize Kiakouama-Maleka, Xavier Quantin, Jean Philippe Spano, Herve Lena, Philippe Fraisse, Henri Janicot, Clarisse Audigier-Valette, Alexandra Langlais, Franck Morin, Alain Makinson, Jacques Cadranel
European Respiratory Journal Aug 2020, 56 (2) 1902066; DOI: 10.1183/13993003.02066-2019
del.icio.us logo Digg logo Reddit logo Technorati logo Twitter logo CiteULike logo Connotea logo Facebook logo Google logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Methods
    • Results
    • Discussion
    • Supplementary material
    • Shareable PDF
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Lung cancer
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

Original Articles

  • Viable virus aerosol propagation by PAP circuit leak
  • Ambulatory management of secondary spontaneous pneumothorax
  • Systematic assessment of respiratory health in illness susceptible athletes
Show more Original Articles

Lung cancer

  • Prognostic factors in non-small cell lung cancer
  • Hyperoxia suppresses lung cancer metastasis through MYC/SLC1A5 pathway
  • Participation in community-based lung cancer screening
Show more Lung cancer

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERJ

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • Podcasts
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN:  0903-1936
Online ISSN: 1399-3003

Copyright © 2023 by the European Respiratory Society